WO2018053019A1 - Compositions et procédés pour le traitement d'infections - Google Patents

Compositions et procédés pour le traitement d'infections Download PDF

Info

Publication number
WO2018053019A1
WO2018053019A1 PCT/US2017/051391 US2017051391W WO2018053019A1 WO 2018053019 A1 WO2018053019 A1 WO 2018053019A1 US 2017051391 W US2017051391 W US 2017051391W WO 2018053019 A1 WO2018053019 A1 WO 2018053019A1
Authority
WO
WIPO (PCT)
Prior art keywords
hwars
enterovirus
infection
medicament
animal
Prior art date
Application number
PCT/US2017/051391
Other languages
English (en)
Inventor
Johnson Yiu-Nam Lau
Philip Man Lung YEUNG
Fuk Woo Jasper CHAN
Manson FOK
Kwok-Yung Yuen
Original Assignee
Ev71 (Hk) Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ev71 (Hk) Limited filed Critical Ev71 (Hk) Limited
Priority to US16/333,114 priority Critical patent/US11826425B2/en
Priority to EP17851466.7A priority patent/EP3512550A4/fr
Priority to CN201780069748.9A priority patent/CN110891603A/zh
Publication of WO2018053019A1 publication Critical patent/WO2018053019A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/42Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum viral
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/50Physical structure
    • C12N2310/53Physical structure partially self-complementary or closed
    • C12N2310/531Stem-loop; Hairpin
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the field of the invention is compositions and methods useful for the treatment of enterovirus infections, particularly enterovirus 71.
  • Enteroviruses are a diverse group of positive-sense single-stranded RNA viruses that are responsible for a number of human and animal diseases, which are typically spread by the fecal- oral route. Such diseases include poliomyelitis, aseptic meningitis, pericarditis/myocarditis, and hand, foot, and mouth disease. A number of enterovirus species are associated with hand, foot, and mouth disease. Some of these (for example, coxsackie virus A 16) produce relatively mild symptoms, whereas other species (notably EV-A71) are associated with the development of severe complications, including brainstem encephalitis, aseptic meningitis, pulmonary edema, and cardio-respiratory collapse.
  • United States Patent Application Publication No. 2014/0023732 provides a combination of traditional Chinese medicines for the treatment of EV-71. These are provided as water or ethanol extracts prepared by soaking of various herbs, then mixed. Unfortunately no data is provided in regards to efficacy of such a combination, and due to variation in the starting materials it is unlikely that a given preparation is reproducible.
  • United States Patent Application No. 2016/0333075 describes the generation of specific monoclonal antibodies directed to EV-71 and, while focusing primarily on diagnostic uses, speculates that such antibodies of sufficiently high affinity could be useful for treatment. The application further describes the development of humanized antibodies having the desired specificity, but notes that such antibodies show tend to show reduced affinity.
  • International Patent Application Publication No. WO 2008/148149 similarly describes passive immunization with polyclonal antibodies derived from eggs or colostrum as a method for treating EV-71 or preventing EV71 infection.
  • Unfortunately wide ranging variation in the amount of virus present in different phases of infection makes determination of the appropriate dosage virus -directed antibodies dependent on accurate staging of the infection. This renders such virus -specific therapeutic antibodies a less than desirable compound for treating viral infection.
  • the inventive subject matter provides compounds and methods that treat and/or prevent enteroviral infection (for example, by EV-A71) and/or their sequelae by reducing accessibility of the enterovirus to hWARS protein on the cell surface.
  • this reduction in accessibility can be accomplished by reducing expression and/or transport of hWARS.
  • this reduction in accessibility can be accomplished by occupancy of hWARS on the cell surface and/or occupancy of enteroviral hWARS receptors that reduce or eliminate interaction between hWARS and the enterovirus.
  • One embodiment of the inventive concept is a method of treating enteroviral (e.g. EV- A71) infection in an animal by introducing an hWARS blocking compound to the animal in an amount effective to at least partially block binding of the enterovirus to hWARS of the animal.
  • the hWARS blocking compound can be an antibody, antibody fragment, or an antibody analog.
  • Such an antibody can be a bispecific antibody having specificity for both hWARS and a co- receptor for the enterovirus (such as hSCARB2 and/or hPSGLl).
  • the hWARS blocking compound can be a small molecule (i.e. having a molecular weight of less than or equal to 800 daltons).
  • the method can include prevention of infection of an animal with the enterovirus.
  • the enterovirus can be EV-A71, or it can be an enterovirus other than EV-A71 (such as Coxsackie virus A16, Coxsackie virus A6, or Coxsackie virus A10).
  • the method can include prevention or treatment of a sequelae of enterovirus infection, such as hand, foot, mouth disease, poliomyelitis, aseptic meningitis, pericarditis, or myocarditis.
  • the method can include passive immunization, wherein the passive immunization provides blocking of a receptor for the enterovirus.
  • the method can include administering a vaccine directed to the enterovirus that provides an active immunization.
  • Another embodiment of the inventive concept is a method of treating infection of an animal by an enterovirus (such as EV-A71) that includes introducing a soluble hWARS or a soluble hWARS analog to the animal in an amount effective to at least partially occupy an hWARS receptor of the enterovirus.
  • the soluble hWARS or the soluble hWARS analog can be an hWARS protein, an hWARS peptide, a chemically modified hWARS protein, a chemically modified hWARS peptide, and/or a small (i.e. having a molecular weight of less than or equal to 800 daltons) molecule analog of hWARS.
  • a chemically modified hWARS protein or chemically modified hWARS peptide can be modified by PEGylation.
  • Such a method can prevent infection of an animal with the enterovirus.
  • the enterovirus can be EV- A71.
  • the enterovirus can be an enterovirus other than EV-A71 (such as Coxsackie virus A16, Coxsackie virus A6, and/or Coxsackie virus A1019).
  • the method of treatment include prevention or treatment of a sequelae of enterovirus infection, such as hand, foot, mouth disease, poliomyelitis, aseptic meningitis, pericarditis, and/or myocarditis.
  • the soluble hWARS or hWARS analog can be co-administered with an hWARS blocking compound.
  • such methods can be used in conjunction with the administration of a vaccine directed to the enterovirus.
  • Another embodiment of the inventive concept is a method of treating infection of an animal by an enterovirus (such as EV-A71) that includes introducing an inhibitor of a proinflammatory peptide (such as IFNy) function to the animal in an amount that is effective to at least partially reduce expression or translocation of hWARS.
  • the inhibition can include reducing expression of the pro-inflammatory peptide, reducing release of the pro-inflammatory peptide, reducing activity of the pro-inflammatory peptide, and/or reducing the activity of a secondary messenger associated with the pro-inflammatory peptide.
  • the inhibitor can be administered in conjunction with soluble hWARS, a soluble hWARS analog, and/or an hWARS blocking compound.
  • the method can used in conjunction with the administration of a vaccine directed to the enterovirus.
  • Another embodiment of the inventive concept is a method of treating infection of an animal by an enterovirus (such as EV-A71) that includes modifying an existing balance between T-helperl and T-helper2 activity in the animal.
  • This balance can be modified by altering (for example, reducing) the activity of a pro-inflammatory peptide, such as IFNy.
  • a pro-inflammatory peptide such as IFNy.
  • Such activity can be reduced by reducing expression of the pro-inflammatory peptide and/or reducing release of the pro-inflammatory peptide.
  • the alteration in activity can be a reduction in the activity of a secondary messenger associated with the pro-inflammatory peptide.
  • an hWARS blocking compound in preparing a medicament useful in treating infection of an animal by an enterovirus (such as EV-A71), where the medicament includes an hWARS blocking compound in an amount that is effective to at least partially block binding of the enterovirus to hWARS.
  • a blocking compound can be an antibody, antibody fragment, an antibody analog, and/or a small molecule (i.e. a molecule having a molecular weight of less than or equal to 800 daltons).
  • the antibody can be a bispecific antibody having specificity for both hWARS and a co-receptor (such as hSCARB2 and hPSGLl) for the enterovirus.
  • Such a medicament can be useful in preventing infection of an animal with an enterovirus.
  • the enterovirus can be EV-A71.
  • the enterovirus can be an enterovirus other than EV-A71, such a Coxsackie virus A16, Coxsackie virus A6, and/or Coxsackie virus A10.
  • the medicament can be useful in preventing or treating sequelae of enterovirus infection, such as hand, foot, mouth disease, poliomyelitis, aseptic meningitis, pericarditis, and myocarditis.
  • the medicament can provide a passive immunization (such as blocking a receptor for the enterovirus), and can include a vaccine directed towards the enterovirus that provides an active immunization.
  • a soluble hWARS and/or a soluble hWARS analog to prepare a medicament useful in treating infection of an animal by an enterovirus (such as EV-A71), where the medicament includes the soluble hWARS and/or soluble hWARS analog in an amount effective to at least partially occupy an hWARS receptor of the enterovirus.
  • the soluble hWARS and/or soluble hWARS analog can be an hWARS protein, an hWARS peptide, a chemically modified hWARS protein, a chemically modified hWARS peptide, and/or a small molecule (i.e.
  • the enterovirus can be EV-A71 or, alternatively, an enterovirus other than EV-A71 (such as Coxsackie virus A16, Coxsackie virus A6, and/or Coxsackie virus A1019).
  • the medicament can be useful in preventing infection of an animal with the enterovirus.
  • the medicament can be useful in preventing and/or treating sequelae of enterovirus infection, such as hand, foot, mouth disease, poliomyelitis, aseptic meningitis, pericarditis, and/or myocarditis.
  • the medicament can include an hWARS blocking compound and/or a vaccine directed to the enterovirus.
  • an inhibitor of a proinflammatory peptide such as IFNy
  • the medicament includes the inhibitor of the pro-inflammatory peptide function in an amount effective to at least partially reduce expression or translocation of hWARS in the animal.
  • Pro-inflammatory peptide function can be reduced by reducing expression of the pro-inflammatory peptide, reducing release of the pro-inflammatory peptide, reducing activity of the pro-inflammatory peptide, and/or reducing activity of a secondary messenger associated with the pro-inflammatory peptide.
  • medicament can include a soluble hWARS, a soluble hWARS analog, an hWARS blocking compound, and/or a vaccine directed to the enterovirus.
  • FIGs. 1A to 1G show data demonstrating a requirement for hWARS activity for EV-A71 infection.
  • FIG. 1A depicts generation of a hWARS knockdown (hWARS KD) cell line.
  • RD cells were stably transfected with shRNA targeting hWARS. The amount of hWARS mRNA was characterized.
  • RD cells transfected with a control shRNA (control RD) are included for comparison.
  • the hWARS KD cells and control RD cells were subsequently challenged with EV-A71.
  • FIG. IB shows the copy numbers of EV-A71 RNAs found in infected hWARS KD cells and control RD cell.
  • FIG. 1A shows generation of a hWARS knockdown (hWARS KD) cell line.
  • RD cells were stably transfected with shRNA targeting hWARS. The amount of hWARS mRNA was characterized.
  • FIG. 1C provides photomicrographs of cells immunostained to show EV-A71 proteins, performed using anti-EV-A71 antibodies.
  • FIG. ID shows typical results of Western blot analyses performed to detect EV-A71 protein in control RD (lane 1) and hWARS KD cells (lane 2) one day after EV-A71 inoculation. Endogenous ⁇ -tubulin was utilized as a loading control.
  • FIG. IE shows viral RNA genome copy number from EV-A71, measured after hWARS KD cells were transfected with hWARS cDNA (lane 1) or an empty vector (lane 2) and followed by challenge with EV-A71.
  • FIG. ID shows typical results of Western blot analyses performed to detect EV-A71 protein in control RD (lane 1) and hWARS KD cells (lane 2) one day after EV-A71 inoculation. Endogenous ⁇ -tubulin was utilized as a loading control.
  • FIG. IE shows viral RNA genome copy number from EV-
  • FIG. 1G shows typical results from characterization of viral loads produced by control RD cells relative to hWARS KD cells.
  • enterovirus receptors for individual serotypes are depicted as follows: "+" indicates receptor usage; "-" indicates receptor not been used; "n.r.” indicates receptor not been reported; EV for enterovirus A; CV-A for coxsackievirus A; E for echovirus; CV-B for coxsackievirus B; PV for poliovirus; EV-D for enterovirus D.
  • CAR for coxsackievirus and adenovirus receptor. Error bars represent the mean + s.d. of three independent experiments. Asterisks indicate significant differences with p ⁇ 0.05. Asterisks with brackets (*) indicate significant differences with p ⁇ 0.05 where the cytopathic effect and the viral load produced in hWARS KD cells were more prominent compared to those of control RD cells. Images shown are representative of three independent experiments.
  • FIGs. 2A to 2H show the impact of hWARS on early EV-A71 replication.
  • FIG. 2A shows the amount of EV-A71 RNA in hWARS-knockdown(KD) RD cells (black line) and control RD (gray line) cells during early infection.
  • FIG. 2B provides photomicrographs of immuno staining for EV-A71 protein in hWARS-KD cells transfected with control RNA (left), hWARS-KD cells transfected with purified EV-A71 RNA (middle), and control RD cells transfected with EV-A71 RNA (right).
  • FIG. 2C shows the results of confocal microscopy for endogenous hWARS expression.
  • FIG. 2D shows results of an attachment assay for EV-A71 to hWARS, hSCARB2 and hPSGL 1.
  • High-titer EV-A71 was incubated with non-permissive L929 cells overexpressing hWARS, hSCARB2 and hPSGL 1, separately, at 4° C for 2 hours, then washed to remove unbound viruses before fixation for immuno staining directed to EV-A71 (left panel) and hWARS, hSCARB2 or hPSGL 1 (middle panel).
  • the degree of co- localization of EV-A71 and hWARS, hSCARB2 or hPSGL 1 was estimated using Mander's Original co-localization coefficient (R).
  • R Mander's Original co-localization coefficient
  • FIG. 2E shows typical results of Western blot analyses of EV-A71 protein in L929 cells transfected with a control vector (lane 1), irrelevant cDNA (lane 2), hWARS-cDNA (lane 3), hSCARB2-cDNA (lane 5) or hPSGL 1-cDNA (lane 6). Mock-transfected L929 (negative control, lane 4) and EV-A71 -infected RD cells (positive control, lane 7) are also shown.
  • FIG. 2F shows results of pull-down studies directed to EV-A71 by recombinant hWARS protein. Recombinant hWARS protein was coupled to agarose before inoculation with three different clinical EV-A71 isolates for overnight at 4° C.
  • FIG. 2G shows the results of anti-hWARS antibodies blockage of EV-A71 infection.
  • Surface hWARS of RD cells were blocked with anti-hWARS antibodies for 1 hour prior to EV- A71 infection.
  • Virus production (TCID 50 ) in conditioned supernatants (top panel) and viral protein expression in infected cell lysates (bottom panel) are shown.
  • FIG. 2H shows saturation of EV-A71 virions by recombinant hWARS protein. EV-A71 was pre-incubated with recombinant hWARS protein prior to challenge of RD cells.
  • Virus production top panel
  • viral protein expression bottom panel
  • r represents the Pearson's correlation coefficient. Error bars in a, g and h represent the mean + s.d. of three independent experiments. Images shown in b, c, d, e, f, g and h are representatives of three independent experiments.
  • FIGs. 3A and 3B show results of hWARS sensitization of cells that are normally non- permissive to EV-A71 infection.
  • FIG. 3A shows IFNy induced hWARS, but not hSCARB2 or hPSGL 1 expression. Different doses of IFNy (0, 10, 50 and 100 U/ml) were administrated to NT2 (left panels) and RD (right panels) cells for 48 hours followed by EV-A71 infection.
  • FIG. 3B shows induced expression and plasma membrane translocation of hWARS after treating with IFNy.
  • NT2 (left panel) and RD (right panel) cells were treated with 100 U/ml IFNy ("+") or mock-treated ("-") for 72 hrs followed by subcellular fractionation.
  • the hWARS protein expression in the cytoplasm and plasma membrane fractions was characterized using anti-hWARS antibodies. Total crude extracts were included as controls. Enrichment of cytosolic protein GAPDH in the cytoplasmic fractions and enrichment of plasma membrane protein sodium potassium ATPase (Na+/K+-ATPase) in the plasma membrane fractions were confirmed by immunoblotting using specific antibodies.
  • FIGs. 4A to 4D show results from EV-A71 infection of mouse cells overexpressing hWARS.
  • FIG. 4A shows typical results of Western blot analyses of EV-A71 protein in EV-A71- challenged neonatal BALB/c mice pre-transduced with empty lentiviral vector (lanes 2 and 3) showing little or mild EV-A71 infection, or lentiviral vector expressing hWARS (lanes 4 and 5) showing positive EV-A71 infection. Two randomly chosen samples were included in each category.
  • FIG. 4B provides representative images of muscle fibers from the hWARS -transduced mice. Tissues of EV-A71 -infected mice were
  • FIG. 4C provides representative images of cells from the brains of the infected (top panels) and uninfected (bottom panels) mice subjected to immunofluorescent staining, with anti-EV-471 antibodies (left panels) or anti-hWARS antibodies (middle panels). Merged images are provided in the right panels.
  • ID shows the results of histopathology studies of EV-A71 -inoculated hWARS -transduced mice. Interstitial infiltration of lymphocytes in various organs were detected using anti-CD 19 antibodies (light gray, left panels). The nuclei were stained with DAPI (dark gray). Hematoxylin and eosin stain showed inflammatory infiltrates of mononuclear cells (arrow heads) and degenerated neuronal cells (arrows) in right panels. Images shown were chosen from three independent experiments.
  • the inventive subject matter provides apparatus, systems and methods in which the finding that tryptophanyl-tRNA synthetase (WARS) can be identified as a cellular surface receptor for enteroviruses, including enterovirus 71 (EV-A71), is utilized in the treatment of EV- 71 infection and in the generation of cell culture and animal models for human EV-A71 infection.
  • WARS tryptophanyl-tRNA synthetase
  • EV-A71 enterovirus 71
  • Compositions and methods that reduce the expression of WARS and/or translocation of WARS to the cell surface, for example by reducing induction by gamma interferon, can reduce entry of EV-A71into susceptible cells.
  • compositions and methods that block WARS at the cell surface can reduce entry of EV-A71into susceptible cells.
  • Such compositions and methods can be useful in the treatment and prevention of infection by EV-A71and other enteroviruses.
  • induction of the expression of WARS in animals and cultured cells can render such animals and cells susceptible to EV-A71 infection, thereby providing non-human models of human disease.
  • animal models and cultured cell models can be utilized in the development of treatment and/or preventative modalities, in addition to aiding in the development of diagnostic tools.
  • inventive subject matter provides many example embodiments of the inventive subject matter. Although each embodiment represents a single combination of inventive elements, the inventive subject matter is considered to include all possible combinations of the disclosed elements. Thus if one embodiment comprises elements A, B, and C, and a second embodiment comprises elements B and D, then the inventive subject matter is also considered to include other remaining combinations of A, B, C, or D, even if not explicitly disclosed.
  • WARS has not been known to act as a viral receptor and has therefore not been utilized as a therapeutic or diagnostic target for enteroviral (EV) infection.
  • Inventors identified WARS as a viral receptor for a variety of EV serotypes following performance of a short-hairpin RNA (shRNA) lentiviral library screening for human genome transcripts that are required for EV-A71 replication. Since direct lentiviral transduction can trigger an
  • Such a screening approach can facilitate the identification of novel immune-inducible cellular factors that are critical for EV-A71 replication.
  • a lentiviral shRNA library targeting 54,509 human transcripts was transduced into 3 x 10 rhabdomyosarcoma (RD) cells, which are highly susceptible to EV-A71 infection.
  • RD cells carrying individual discrete shRNAs were challenged using a high titer of EV-A71.
  • the inventors theorize that knockdown of a cellular gene that is critical for EV-A71 replication would halt the virus replication and hence protect the cells from EV-A71 -induced cytopathic effect (CPE).
  • CPE cytopathic effect
  • Inventors identified 118 candidate genes, the knockdown of which protected RD cells from EV-A71 -induced CPE (see GEO accession number: GSE80407).
  • Candidate genes were selected for validation based on their expression levels and the number of unique shRNAs targeting the same gene. Without wishing to be bound by theory, Inventors anticipate that knockdown of highly expressed genes could have a more pronounced effect on EV- A71 replication, and that knockdown by multiple unique shRNAs would prevent off- targeting. Filtering by these criteria led to a candidate human gene named hWARS, silencing of which effectively protected RD cells from EV-A71 -induced CPE.
  • FIG. 1A To establish the importance of hWARS in EV-A71 replication, an independent RD cell clone stably expressing hWARS shRNA was generated (see FIG. 1A). This clone was consistently found to be highly resistant to EV-A71 infection (see FIG. IB, 1C, and ID).
  • Enterovirus B serotypes such as Echovirus 6, 11, 25, 30 and Coxsackie B2 and B5 are especially known to cause infantile liver failure, myocarditis, pericarditis, pneumonia, encephalitis, and sudden or cot death.
  • a major 2014 US outbreak of Enterovirus D68 was associated with severe respiratory illness and fatal acute flaccid myelitis. It should be appreciated that different cellular receptor for host cell entry can be used by different serotypes of enteroviruses within the same species group.
  • hWARS was initially identified as an aminoacyl-tRNA synthetase catalyzing the attachment of tryptophan onto cognate tRNA, additional roles in angiogenesis, cytoskeletal reorganization and shear stress-responsive gene expression have also been suggested. Its function as an enterovirus receptor protein, however, has not been previously known.
  • EV-A71 has a positive-stranded RNA genome that can serve as template for the synthesis of both viral proteins and RNAs. Inventors reasoned that both processes would be affected if hWARS was a key functional element at an early stage of the infection process (e.g. prior to the bifurcation of viral RNA replication and viral protein synthesis). Both viral proteins and viral RNAs were found to be significantly reduced in hWARS -knockdown RD cells (FIG. IB, 1C and 1G), indicating that a hWARS is involved in a early stage of the EV-A71 infective process.
  • EV-A71 pathogenesis has generally been attributed to the tissue expression patterns of two known cellular receptors: human scavenger receptor class B member 2 (hSCARB2) and human P-selectin glycoprotein ligand-1 (hPSGLl).
  • hSCARB2 human scavenger receptor class B member 2
  • hPSGLl human P-selectin glycoprotein ligand-1
  • Inventors also compared the EV-A71 infection efficiency in L929 cells expressing hWARS, hSCARB2, and hPSGLl.
  • Such antibodies, antibody fragments, and/or antibody can be provided as an intravenous infusion, for example as a solution or suspension in a saline solution.
  • hWARS-specific antibodies, antibody fragments, and/or antibody analogs can be provided in lyophilized or dried form that is reconstituted prior to use.
  • EV-A71 (or other suitable enterovirus serotype) infection anti-hWARS antibody, antibody fragment, and/or antibody analog can be supplied at from 10 ⁇ g/kg to 10 mg/kg body weight.
  • a single infusion of antibody, antibody fragment, and/or antibody analog can be effective.
  • repeated or continuous infusion can be used.
  • hWARS-specific antibodies, antibody fragments, and/or antibody analogs can be provided in combination with one or more anti-viral and/or anti-inflammatory compounds.
  • anti-viral and/or anti-inflammatory compounds can be included with an hWARS- specific antibody, antibody fragment, and/or antibody analog containing composition, or can be co-administered. If co-administered the anti- viral and/or anti-inflammatory
  • hWARS-specific antibody, antibody fragment, and/or antibody analog composition can be provided on the same schedule as the hWARS-specific antibody, antibody fragment, and/or antibody analog composition, or on a different schedule.
  • hWARS proteins, peptides, and/or analogs thereof can be used to treat and/or prevent enterovirus infections.
  • hWARS proteins, peptides, and/or analogs can be provided as an intravenous infusion, for example as a solution or suspension in a saline solution.
  • hWARS proteins, peptides, and/or analogs can be provided in lyophilized or dried form that is
  • hWARS proteins, peptides, and/or analogs can be supplied at from 10 ⁇ g/kg to 10 mg/kg body weight. In some embodiments a single infusion of hWARS proteins, peptides, and/or analogs can be effective. In other embodiments repeated or continuous infusion can be used. Repeated infusions can be applied at intervals ranging from 3 times a day to once a week.
  • hWARS proteins, peptides, and/or analogs can be provided in combination with one or more anti-viral and/or anti-inflammatory compounds.
  • anti-viral and/or anti-inflammatory compounds can be included with an hWARS proteins, peptides, and/or analogs containing composition, or can be co-administered. If co-administered the anti- viral and/or anti-inflammatory compounds can be provided on the same schedule as the hWARS proteins, peptides, and/or analogs composition, or on a different schedule.
  • hWARS represents a novel EV-A71 cellular receptor that provides opportunities for treatment modalities that can reduce or eliminate enterovirus infections, for example by reducing expression of hWARS, blocking hWARS at the cell surface (for example, with specific antibodies, antibody fragments, and/or antibody analogs), and/or saturating viral binding sites with hWARS protein, peptides, and/or analogs (for example, small molecule analogs). It is known that EV-A71 can enter host cells using different cellular receptors.
  • hSCARB2 Due to the relatively robust expression of hSCARB2 in lysosomes and endosomes of many cell types, hSCARB2 has been proposed to be the major receptor for systemic EV-A71 infection. Its expression pattern, however, cannot fully explain clinical complications, especially in the cases of severe EV-A71 infection.
  • the identification of hPSGLl, a leukocyte-specific membrane protein as an alternative receptor for EV-A71 infection has been proposed for the infiltration of EV-A71 -infected leukocyte into the central nervous system which accounts for the neurotropism of EV-A71.
  • the absolute requirement of hPSGLl for EV-A71 cell entry remains elusive as not all tested EV-A71 strains can utilize this receptor for infection. It remains unclear which cellular receptor plays a key role in EV- A71 neurotropism.
  • hWARS expression can respond to external stimulation by IFNy (FIG. 3A). Since cytokine response is a common mechanism for anti-viral defense, without wishing to be bound by theory Inventors believe that certain cell types can become sensitized to EV-A71 infection following such a host-mounted innate immune response to primary infection. This was tested using a human teratocarcinoma cell line, NT2, which exhibits properties of a committed neuronal precursor at an early stage of development. Under low-titer infection conditions, NT2 can only moderately support EV-A71 replication (FIG. 3 A, lane 1; left).
  • IFNy was then added to the culture media to mimic the cytokine production that can occur upon primary infection.
  • specific antibodies and primers for Western blot and quantitative reverse transcription polymerase chain reaction (qPCR) analyses respectively, the inventors identified an enhanced expression of hWARS (but not hSCARB2 or hPSGLl) in cells treated with IFNy, in a dose-dependent manner (FIGs. 3A and 3C).
  • hWARS plasma membrane translocation of hWARS was also observed upon IFNy stimulation (FIG. 3B). This is consistent with an infection model where hWARS can act as an IFNy-inducible cellular receptor for EV-A71 infection.
  • IFNy inhibition can be provided by the administration of an antibody, antibody fragment, and/or antibody analog specific for IFNy.
  • the functions of IFNy can be inhibited, for example by administration of glucocorticoids (such as dexamethasone), short chain fatty acids (such as butyrate), and certain proteins (such as CC16).
  • glucocorticoids such as dexamethasone
  • short chain fatty acids such as butyrate
  • proteins such as CC16
  • Inventors contemplate that administration of such proteins and/or compounds can provide an effective therapeutic mode for treatment of infection by enteroviruses (such as EV-A71) and/or their sequelae.
  • Such proteins or compounds can be administered in combination with antibodies, antibody fragments, and/or antibody analogs directed towards hWARS or in combination with hWARS proteins, peptides, and/or analogs in the treatment of infection by enteroviruses (such as EV-A71) and/or their sequelae.
  • enteroviruses such as EV-A71
  • hWARS can act as a functional receptor for EV-A71 infection in vivo
  • the inventors developed a mouse model which overexpressed hWARS by a lentiviral vector (lenti-) expression system. Successful gene delivery by such a mechanism was first confirmed by the detection of hWARS expression in lenti-hWARS -transduced L929 cells. Subsequently Inventors transduced neonatal BALB/c mice with 10 6 copies of lenti-hWARS (using empty lentiviral vector as a control) via intraperitoneal, intracerebral, and subcutaneous injection simultaneously. hWARS was allowed to be overexpressed for 5 days before the mice were challenged with EV-A71.
  • lenti- lentiviral vector
  • Inventors detected EV-A71 proteins and RNAs in skeletal muscles and brains of mice transduced with lenti- hWARS (FIG. 4A, 4B, and 4C). In contrast, viral protein and/or RNA was barely detectable or not detected in control mice transduced with empty lentiviral vector followed by EV- A71 inoculation.
  • Pathologically Inventors observed interstitial infiltration of inflammatory cells in various organs (FIG. 4D; left panels). The inventors also observed degeneration of neurons, which was consistent with the ataxia and paralysis observed with the EV-A71 -inoculated lenti- hWARS -transduced mice.
  • Such a mouse model can fully or partially recapitulate the neurological symptoms associated with EV-A71 infection in humans.
  • Such a mouse model can serve as a research tool in the development of safe and effective therapies for enterovirus infection and/or prevention or treatment of neurological sequelae of enteroviral infection, and can also support the development of diagnostic tools.
  • Proinflammatory cytokines induced by early innate immune response often help to control virus replication and prime humoral and cell-based adaptive immune responses.
  • a subset of proinflammatory cytokines, including IFNy is significantly elevated in patients with severe EV-A71 infections.
  • hWARS novel EV-A71 cellular receptor
  • hWARS novel EV-A71 cellular receptor
  • Blocking the accessibility of hWARS by specific antibodies or recombinant proteins (as noted above) can be potential treatment options for the EV-A71-associated severe neurological complications.
  • medicaments that incorporate such proteins, peptides, and/or compounds can be useful in treating or preventing enterovirus (for example, EV-A71) infection and/or its sequelae.
  • T-helperl activity and T-helper2 activity can play an important and previously undocumented role in the control of viral infection, and that the effectiveness of reducing IFNy activity in reducing or eliminating enteroviral infection is due at least in part to alteration of this balance.
  • Alteration of the balance between T-helperl and T- helper2 activity can, therefore, provide a method for treating or preventing an enterovirus (for example, EV-A71) infection and/or sequelae of such an infection. This can, for example, be accomplished using compounds that interact with toll-like receptors, treatment with
  • glucocorticoids and treatment with CpG oligodeoxynucleotides, treatment with calcitriol, and/or treatment with other immune modulators.
  • Inventors believe that use of medicaments that incorporate such compounds can be useful in treating or preventing enteroviral infection and/or its sequelae.
  • each of the above described treatment modalities can be combined with one another in order to provide a combined and/or synergistic effect.
  • such treatment modalities can be used in conjunction with the administration of a preventative or therapeutic vaccine and/or antiviral compound directed to an enterovirus.
  • NT2 ATCC
  • RD ATCC
  • FCS heat-inactivated fetal calf serum
  • mice Female BALB/c mice at 5 days of age were obtained from the Laboratory Animal Unit, the University of Hong Kong. The animals were housed in SPF-free facilities with 12 hours light-dark cycles together with lactating mothers. All animal-related experiments were performed according to the standard operating procedures as we previously described26 and approved by the University of Hong Kong Committee on the Use of Live Animals in Teaching and Research.
  • FMV-based shRNA library Feline immunodeficiency virus (FIV)- based shRNA library was produced. Briefly, 2 ⁇ g of the shRNA library in lentiviral constructs (SBI) were co-transfected with 10 ⁇ g of the pPACK packaging plasmid mix into HEK293T cells using Lipofectamine and Plus reagents according to the manufacturer's protocol (Invitrogen). At 24 and 72 hours after transfection, conditioned culture media were collected to harvest the packaged viruses. In parallel, GFP-packaged viruses were produced. The infectivity of the viruses was established based on the percentage of the GFP-transduced RD cells.
  • shRNA-expressing vector constructs (Mission shRNA) were purchased from Sigma. These shRNA-packaged viruses were generated in HEK293T cells using ViraPowerTM Lentiviral Expression Systems (Invitrogen) according to the manufacturer's protocol.
  • RNA isolation kit (Ambion). Five microgram of total RNAs were reverse transcribed into cDNAs in the presence of 10 ⁇ of cDNA synthesis GNF primer (5'- ATTTATTGTATCTGTGGGAGCCTC-3 ' ) (SEQ ID NO. 1),100 mM dithiothreitol, 10 mM of each dNTP, IX reverse transcriptase buffer, and 200 U of Superscript® III Reverse
  • PCR amplification of the shRNA target region was performed under the following conditions: Step 1: 94° C for 4 minutes; Step 2: 94° C for 30 second and then 68° C for 1 minute; Step 3: Repeat Step 2 for 20 cycles; Step 4: 68° C for 3 minutes. Aliquots were made by transferring 1 ⁇ of the first round PCR products into 4 new tubes, each containing 1 X PCR reaction buffer, 20 mM of dNTP, 20 ⁇ each of NRev GNF universal primer (5'- AAAGAATGCTTATGGACGCTAGAA-3 ' ) (SEQ ID NO. 4) and NFwd-Bio primer (Biotin-5'- CTTCCTGTC AGA-3 ' ) (SEQ ID NO.
  • PCR for biotin- labeling was performed under the following conditions: Step 1: 94° C for 2 minutes, 50° C for 2 minutes and then 68° C for 1 minute; Step 2: 94° C for 30 second and then 68° C for 30 second; Step 3: Repeat Step 2 for 18 cycles; Step 4: 68° C for 3 minutes.
  • the PCR products were then gel purified using QIAquick PCR purification kit (QIAGEN) as described in the manufacturer's protocol. After purification, the PCR product was treated with Lambda exonuclease (NEB) at 37° C for 2 hours to remove the non-biotinylated strand.
  • QIAGEN QIAquick PCR purification kit
  • the digested products were purified as described above and then quantified. Ten microgram of the purified products were hybridized on the GeneChip® Human Genome U133+2 Array (Affymetrix) using the Affymetrix hybridization buffer followed by staining with streptavidin-phycoerythrin
  • RNAs were isolated using mirVana miRNA isolation kit(Ambion). The RNAs were first quantified by NanoDrop® Spectrophotometer (ND-1000; Thermo Scientific). One microgram of the total RNAs were reverse transcribed as described above. qPCR was performed using FS Universal SYBR Green MasterRox (Roche) reaction mix with the temp-cycling condition of 15 second at 95° C followed by 1 minute at 60° C for 55 cycles in a 7900 Real Time PCR System (Applied Biosystems).
  • EV- A71 -infected samples' cDNAs were quantified using 300 nM each of forward and reverse specific primers (5'-CCCCTGAATGCGGCTAATCC-3 ⁇ SEQ ID NO. 6, and 5'- ACACGGACACCC AAAGTAGT-3 ' , SEQ ID NO. 7).
  • forward and reverse specific primers (5'- GCCCCTGAATGCGGCTAAT-3', SEQ ID NO. 8, and 5 ' - ATTGTCACCATAAGCAGYCA-3 ' , SEQ ID NO. 9) and a probe (5'FAM-
  • Housekeeping gene GAPDH expression was also measured as an internal control using primers 5 ' -TC ACC ACC ATGGAGAAGGC-3 ' (SEQ ID NO. 11) and 5'-
  • Hs00188259_ml After reverse transcription, the cDNAs were added to a reaction mixture containing the TaqMan universal PCR master mix (Applied Biosystems) and the TaqMan gene expression assay mix (inventoried for the corresponding genes; Applied Biosystems).
  • Quantitative PCRs were carried out using the same conditions described above.
  • knockdown cells were fixed in 4% paraformaldehyde with or without 0.1 % triton X-100. After 1 hour blocking with 3% bovine serum albumin (BSA) at room temperature, the cells were stained with anti-EV- A71 antibodies (Millipore) and/or with anti-hWARS antibodies (Abeam) for 1 hour at room temperature as we previously described.30 Unbound antibodies were washed away 6 times with PBS. Positively stained cells were detected by secondary IgG (H+L) antibodies conjugated either with Alexa Fluor 488 or Alexa Fluor 594 (Life Technology) for 30 minutes at room temperature.
  • BSA bovine serum albumin
  • the stained cells were mounted onto glass slides with VECTASHIELD mounting medium with 4', 6'-diamidino-2- phenylindole (DAPI) (Vector Lab) and examined with a Leica TCS-NT microscope (Leica Microsystem) or a LSM700 confocal microscope (Zeiss).
  • DAPI 6'-diamidino-2- phenylindole
  • the tissue sections were first de-paraffinized and rehydrated, followed by treatment with Antigen Unmasking Solution (Vector Lab) to retrieve the antigens according the manufacturer's instructions. Detection of antigens was done under the same conditions as described above.
  • the membranes were stripped with Restore Western blot stripping buffer (Pierce) before reprobing with anti-y-tubulin antibodies (Sigma), anti-Na+/K+-ATPase antibodies (Abeam) and anti-GAPDH antibodies (Abeam).
  • agarose-bound proteins were fractionated by SDS-PAGE and detected by Western blot analysis using anti-hWARS antibodies (Abeam) and anti-EV-A71 antibodies (Millipore).
  • Subcellular fractionation Subcellular fractionation of RD and NT2 cells were performed using the Plasma Membrane Protein Extraction Kit (Abeam) following the
  • the cytoplasmic fraction (supernatant) and the total cellular membrane (pellet) fractions were further separated by centrifugation at 10,000 x g for 30 minutes at 4° C.
  • Plasma membrane proteins were further extracted by resuspending the total cellular membrane pellet in 200 ⁇ of the Upper Phase Solution and 200 ⁇ of the Lower Phase Solution.
  • the complex was then incubated on ice for 5 minutes before centrifugation at 1000 x g for 5 minutes at 4° C.
  • the upper phase was collected and the steps were repeated by adding 100 ⁇ Lower phase solution.
  • the upper phase was collected and combined with the previously harvested upper phase.
  • the resultant was then diluted in 5 volumes of water and kept on ice for 5 minutes.
  • the plasma membrane proteins were collected by centrifugation of the diluted upper phase at 10,000 x g for 10 minutes at 4° C.
  • 0.5 % Triton X-100 in PBS was added to dissolve the plasma membrane protein pellets for Western blot analysis.
  • ViraPowerTM Lentiviral Expression Systems (Invitrogen) as described above.
  • mice from each group were sacrificed to harvest their heart, brain, muscles, kidney, lung, liver and spleen, respectively, at day 5 after virus inoculation. Four independent experiments were done. The collected organs were divided into different sets for qPCR, Western blot, immunohistochemical and histopathological analyses. [0067] It should be apparent to those skilled in the art that many more modifications besides those already described are possible without departing from the inventive concepts herein. The inventive subject matter, therefore, is not to be restricted except in the spirit of the appended claims. Moreover, in interpreting both the specification and the claims, all terms should be interpreted in the broadest possible manner consistent with the context.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Virology (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Oncology (AREA)
  • Biochemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Communicable Diseases (AREA)
  • Genetics & Genomics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biophysics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Microbiology (AREA)
  • Engineering & Computer Science (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Il a été découvert que des entérovirus, tels qu'EV-A71, utilisent hWARS en tant que récepteur de surface cellulaire, l'expression de hWARS rendant les cellules susceptibles d'infection entérovirale. La réduction de l'activité de surface cellulaire hWARS réduit la susceptibilité d'infection entérovirale et fournit un mode pour le traitement ou la prévention d'une infection entérovirale et de ses séquelles. De manière similaire, l'expression de hWARS dans des cellules cultivées et des modèles animaux fournit des modèles pour comprendre des mécanismes de maladie entérovirale et le développement de vaccins et/ou de produits pharmaceutiques pour prévenir ou traiter une maladie entérovirale.
PCT/US2017/051391 2016-09-13 2017-09-13 Compositions et procédés pour le traitement d'infections WO2018053019A1 (fr)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US16/333,114 US11826425B2 (en) 2016-09-13 2017-09-13 Compositions and methods for treatment of enterovirus infection
EP17851466.7A EP3512550A4 (fr) 2016-09-13 2017-09-13 Compositions et procédés pour le traitement d'infections
CN201780069748.9A CN110891603A (zh) 2016-09-13 2017-09-13 用于治疗肠道病毒感染的组合物和方法

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201662394061P 2016-09-13 2016-09-13
US62/394,061 2016-09-13

Publications (1)

Publication Number Publication Date
WO2018053019A1 true WO2018053019A1 (fr) 2018-03-22

Family

ID=61619237

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2017/051391 WO2018053019A1 (fr) 2016-09-13 2017-09-13 Compositions et procédés pour le traitement d'infections

Country Status (4)

Country Link
US (1) US11826425B2 (fr)
EP (1) EP3512550A4 (fr)
CN (1) CN110891603A (fr)
WO (1) WO2018053019A1 (fr)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3512550A4 (fr) * 2016-09-13 2020-05-06 EV71 (HK) Limited Compositions et procédés pour le traitement d'infections

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016137280A1 (fr) * 2015-02-26 2016-09-01 재단법인 의약바이오컨버젼스연구단 Composition pour le traitement ou la prévention de maladies infectieuses inflammatoires ou composition pour l'amélioration de l'immunité comprenant de la tryptophanyl-arnt synthétase en tant que principe actif

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008148149A1 (fr) 2007-06-05 2008-12-11 Anadis Ltd Procédé de traitement ou d'inhibition des infections à entérovirus 71
US8263570B2 (en) 2008-12-15 2012-09-11 National Taiwan University Treating picornavirus infection by targeting MicroRNA miR-141
EP2939689B1 (fr) * 2009-12-11 2017-09-13 aTyr Pharma, Inc. Glutaminyl ARNt synthétases pour leur utilisation dans le traitement de maladies inflammatoires
WO2012060779A1 (fr) 2010-11-02 2012-05-10 Singapore Polytechnic Méthode de détection de l'entérovirus ev71
CN102716245B (zh) 2011-03-31 2014-05-28 丽珠医药集团股份有限公司 中药组合物在制备抗71型肠道病毒的药物中的用途
CN103841993A (zh) 2011-08-26 2014-06-04 淡马锡生命科学实验室有限公司 人肠道病毒特异性抗体及其在诊断中的应用
TWI700295B (zh) 2013-12-16 2020-08-01 馬來西亞商Mab探索私人有限公司 對會感染人類之腸病毒具有專一性之抗體
EP3512550A4 (fr) * 2016-09-13 2020-05-06 EV71 (HK) Limited Compositions et procédés pour le traitement d'infections

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016137280A1 (fr) * 2015-02-26 2016-09-01 재단법인 의약바이오컨버젼스연구단 Composition pour le traitement ou la prévention de maladies infectieuses inflammatoires ou composition pour l'amélioration de l'immunité comprenant de la tryptophanyl-arnt synthétase en tant que principe actif

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
ANDREEV, D. E. ET AL.: "Glycyl-tRNA synthetase specifically binds to the poliovirus IRES to activate translation initiation", NUCLEIC ACIDS RESEARCH, vol. 40, no. 12, 2012, pages 5602 - 5614, XP055498305 *
GUO, F. ET AL.: "Specific inhibition of the synthesis of human lysyl-tRNA synthetase results in decreases in tRNALys incorporation, tRNA3Lys annealing to viral RNA, and viral infectivity in human immunodeficiency virus type 1", JOURNAL OF VIROLOGY, vol. 77, no. 18, 2003, pages 9817 - 9822, XP055498310 *
HURDLE, J. G. ET AL.: "Prospects for aminoacyl-tRNA synthetase inhibitors as new antimicrobial agents", ANTIMICROBIAL AGENTS AND CHEMOTHERAPY, vol. 49, no. 12, 2005, pages 4821 - 4833, XP002626387 *
LEE, C.-W. ET AL.: "Overexpressed tryptophanyl-tRNA synthetase, an angiostatic protein, enhances oral cancer cell invasiveness", ONCOTARGET, vol. 6, no. 26, 2015, pages 21979 - 21992, XP055498307 *
See also references of EP3512550A4 *

Also Published As

Publication number Publication date
EP3512550A1 (fr) 2019-07-24
EP3512550A4 (fr) 2020-05-06
US20190358326A1 (en) 2019-11-28
US11826425B2 (en) 2023-11-28
CN110891603A (zh) 2020-03-17

Similar Documents

Publication Publication Date Title
Bjelobaba et al. Multiple sclerosis and neuroinflammation: the overview of current and prospective therapies
Pan et al. MS2 VLP-based delivery of microRNA-146a inhibits autoantibody production in lupus-prone mice
JP2021138721A (ja) Hiv予備免疫化および免疫療法
Zhang et al. miR-21a-5p promotes inflammation following traumatic spinal cord injury through upregulation of neurotoxic reactive astrocyte (A1) polarization by inhibiting the CNTF/STAT3/Nkrf pathway
CN114025785B (zh) 慢性炎症和病毒感染的诊断和治疗
MX2015002491A (es) Metodos para el tratamiento de infecciones de hepatitis b y hepatitis d.
Rajalakshmy et al. Hepatitis C virus NS3 mediated microglial inflammation via TLR2/TLR6 MyD88/NF-κB pathway and toll like receptor ligand treatment furnished immune tolerance
Nath et al. Novel approaches and challenges to treatment of central nervous system viral infections
Su et al. The heat shock protein 70 family of chaperones regulates all phases of the enterovirus A71 life cycle
Zhang et al. An RNA Interference/Adeno‐Associated Virus Vector–Based Combinatorial Gene Therapy Approach Against Hepatitis E Virus
US11826425B2 (en) Compositions and methods for treatment of enterovirus infection
Tan et al. Inhibition of enterovirus 71 infection by antisense octaguanidinium dendrimer-conjugated morpholino oligomers
TWI670064B (zh) 抗病毒劑及治療病毒感染之方法
EP2688574B1 (fr) Méthode de criblage de composés antirétroviraux et vaccin
US20220220483A1 (en) Method for inhibiting infection and activation of virus
Lalani et al. Molecular mechanism of L-SP40 peptide and in vivo efficacy against EV-A71 in neonatal mice
RU2526146C2 (ru) Способ профилактики и лечения бронхиальной астмы, осложняющих ее респираторных вирусных инфекций и других воспалительных заболеваний дыхательных путей
Baharlooi et al. Nucleic acids as novel therapeutic modalities to address multiple sclerosis onset and progression
AU2019373090B2 (en) Compositions and methods for treating viral infections
AU2014285701B2 (en) RNAi agent for inhibition of Chikungunya virus
Manna et al. Therapeutic Agents for COVID-19: an Overview
Ge et al. May early intervention with intravenous immunoglobulin pose a potentially successful treatment for Ebola virus infection?
WO2012030778A1 (fr) Séquences de kobuvirus canins et leurs utilisations
US20230000917A1 (en) Treatment of hepatitis b virus (hbv) infection
US20170130229A1 (en) Methods and compositions for inhibiting infection by influenza and viruses

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 17851466

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2017851466

Country of ref document: EP

Effective date: 20190415